• Open Access
    Review

    Potential role of resveratrol and its nano-formulation as anti-cancer agent

    Akshay Kumar 1
    Balak Das Kurmi 2
    Amrinder Singh 2
    Dilpreet Singh 2*

    Explor Target Antitumor Ther. 2022;3:643–658 DOI: https://doi.org/10.37349/etat.2022.00105

    Received: August 01, 2022 Accepted: August 22, 2022 Published: October 31, 2022

    Academic Editor: Katrin Sak, NGO Praeventio, Estonia

    This article belongs to the special issue Plant Extracts as an Infinite Resource for New Anticancer Agents

    Abstract

    The uncontrolled and metastatic nature of cancer makes it worse and more unpredictable. Hence, many therapy and medication are used to control and treat cancer. However, apart from this, many medications cause various side effects. In America, nearly 8% of patients admitted to the hospital are due to side effects. Cancer is more seen in people residing in developed countries related of their lifestyle. There are various phytoconstituents molecules in which resveratrol (RSV) is the best-fitted molecule for cancer due to its significantly less adverse effect on the body. RSV inhibits the initiation and progression of cell proliferation due to the modulation of various pathways like the phosphoinositol 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway. RSV downgraded cell cycle-regulated proteins like cyclin E, cyclin D1, and proliferating cell nuclear antigen (PCNA) and induced the release of cytochrome c from the mitochondria, causing apoptosis or programmed cell death (PCD). A great benefit comes with some challenges, hence, RSV does suffer from poor solubility in water i.e. 0.05 mg/mL. It suffers from poor bioavailability due to being highly metabolized by the liver and intestine. Surprisingly, RSV metabolites also induce the metabolism of RSV. Hence, significantly less amount of RSV presented in the urine in the unchanged form. Due to some challenges like poor bioavailability, less aqueous solubility, and retention time in the body, researchers concluded to make the nanocarriers for better delivery. Adopting the technique of nano-formulations, increased topical penetration by up to 21%, improved nano-encapsulation and consequently improved bioavailability and permeability by many folds. Hence, the present review describes the complete profile of RSV and its nano-formulations for improving anti-cancer activity along with a patent survey.

    Keywords

    Resveratrol, cancer, solubility, nano-formulations, synergism

    Introduction

    Cancer is a genetic disease caused by various factors like DNA mutation, including radiation that may be ultraviolet radiation, and some environmental factors, alteration in cell division [1]. Cancer is more predominant in developed countries where their living standards are high, like the United States of America (USA), where approximately 1.9 million patients are diagnosed with cancer, and about 609,360 deaths of cancer in 2022 [2]. The most common cancer is in the lungs, prostate, and colorectal cancer in about 43% of total cancer in males. However, in females, the predominant cancer is breast, lung, and colorectal in about 50% of total cancer in female surveys in 2020, according to the national cancer institute, USA. The mortality due to cancer is higher in males (189.5 per 100,000) than in women (135.7 per 100,000) [3]. Cancer treatment contains one or a combination of methods used to treat or minimize the severity of symptoms like biomarker testing for cancer treatment, chemotherapy, hormone therapy, radiation therapy, immunotherapy, and surgery. Natural drugs benefit from synthetic drugs because they have fewer side effects, especially when cancer is concerned. In America, 8% of patients are admitted to hospitals due to the side effects of synthetic drugs [4].

    Resveratrol (3,5,4’-trihydroxy-trans-stilbene, RSV) is obtained from various plant-based sources and also obtained from synthesis and recombinant technology. It was first isolated in 1939 from Veratrum grandiflorum O. Loes [5]. RSV was found to manage various disease conditions due to its binding to different enzymes and proteins of various pathways, which regulate the gene expressions or modulate the activity of other processes necessary for its effect. Apart from all its beneficial values for human health, it has some challenges, including extensive bioavailability and metabolism by the enzymes in the liver and intestine and undergoing phase 2 metabolism [6]. RSV has very effective properties to have an anti-cancer agent [7]. It also has other properties, which are discussed in Figure 1. Nanocarriers have advantages when it comes to delivering the drug to the targeted site or, in general, with improved bioavailability and therapeutic efficiency of the drug due to the accumulation of the drug at a particular site [8]. Nanocarriers comprise various nanomaterials that transport chemicals [active pharmaceutical ingredient (API)] to various body parts. Nanocarriers enable fascinating advantages over the conventional delivery system. This system increases in vivo efficiency by targeting drug delivery to the specific site, such as the cellular level due to the targeting ligand attached to the surface to nanocarriers, favorable biodistribution, enhanced intracellular penetration, and drug delivery circulates a long time in blood [9]. All these characteristics make the nanocarriers a perfect candidate for chemotherapy. Nanoformulation plays a crucial role. In this case, RSV has very low solubility and bioavailability which the nanoformulation improves to some extent. Hence, the present review gives a brief art of RSV as an anti-cancer agent, its associated formulations for improving therapeutic activity, and a patent survey of new technologies.

    Chemical structure of RSV in the trans and cis form

    RSV: an overview

    RSV is obtained from natural resources of red grapes, blueberries, raspberries, mulberries, cranberries, red wine, nuts, and peanuts [10]. RSV is extracted from wild Polygonum cuspidatum’s root [11]. RSV can also synthesize in the laboratory by the Witting reaction and decarbonylative Heck reaction [12]. RSV occurs in two forms, cis-RSV and trans-RSV (Figure 1), in which trans-isoforms are easily converted into cis-isoforms (cis-RSV) by heat and ultraviolet radiation [13]. Using recombinant technology, RSV is produced by yeast (Saccharomyces cerevisiae) with the help of glucose and ethanol, which is used in the cosmetic ingredients and food supplement industry [14]. It is a chemical stilbenoid (polyphenolic, phytoalexin) released by various plants during stress or injury during a pathogen attack (bacteria, fungi) [15]. Moreover, trans-RSV effectively managed cognitive disorders like attention deficit hyperactivity disorder (ADHD), Alzheimer’s, and dementia [16].

    RSV has the chemical formula: C14H12O3, molecular weight: 228.24, melting point: 254, boiling point: 449.1 ± 14, Log P: 3.10 and solubility in water is 0.05 mg/mL [17] and in polyethylene glycol (PEG) 400 is about 374 mg/mL [18]. RSV consists of phytoestrogen similar to estradiol, a major constituent of estrogen. RSV caused alteration in almost 127 pathways, polyacrylamide gel electrophoresis (PAGE) analysis says. It includes glycolysis, Krab’s cycle [tricarboxylic acid (TCA) cycle], electron transport chain, oxidative phosphorylation, insulin signaling, and sterol biosynthesis. RSV protects and enhances living standards by having anti-cancer, anti-oxidant, anti-inflammatory, and neuroprotective properties [19]. RSV is highly lipophilic. It comes under biopharmaceutical classification system 2 (BCS2) [20]. It can cross the membrane and act on the brain by entering the blood-brain barrier (BBB) [21]. Due to its low pharmacokinetic properties, such as solubility in an aqueous medium (0.05 mg/mL), highly metabolized and excreted by the body, it makes highly inefficient oral doses. These doses form overall have very low bioavailability [22]. Oral absorption of RSV is relatively high, about 75%, but the drug rapidly metabolizes through first-pass metabolism by the liver and intestine [23]. Due to this concern about the pharmacokinetic improvement of doses form, increase in demand. Nanotechnology plays a significant role in delivering formulation to the demands of pharmacokinetic profiles [24]. Many in vitro studies and animal studies found that RSV has the potential to exert a favorable effect on clinical studies [25]. RSV targets various enzymes and proteins such as cyclooxygenases (COXs), lipoxygenases, kinases, sirtuins (SIRTs), ribonucleotide reductase, and DNA polymerase to show biological action [26]. The major activities of RSV are given in Figure 2.

    Biological effects of RSV

    RSV: biopharmaceutical challenges associated with different routes of administration

    There are many challenges with RSV delivery using various routes. RSV possesses low solubility in an aqueous medium and almost zero bioavailability profile which is not fit for oral and topical applications alone. That is where novel drug delivery systems play an essential role. Although it is a perfect anti-oxidant, chemoprotective and anti-inflammatory agent [27], problems arise due to insolubility, less skin penetration, poor photostability, and bioavailability.

    Oral route—RSV contributes various effects, as discussed above, but comes with challenging problems like low solubility (in water—0.05 mg/mL) and metabolism [17]. RSV quickly absorbs into the portal veins nearly 75% due to its lipophilic nature having Log P/Ko/w is 3.10. On the other hand, it is aggressively metabolized by the liver through phase 2 metabolism and the formation of sulfated, methylated, and glucuronidated complexes. Sulfate complexes with phenolic groups play a rate-limiting step in the bioavailability of RSV. Some of these complexes come back to the gastrointestinal tract (GIT) to induce metabolism [6]. It is fascinating to know that RSV itself increases the metabolism of its own. A significantly less amount of drug passes to the systemic circulation in unchanged form, but most drugs make complexes with albumin and lipoprotein. It also enabled passive diffusion and temperature-mediated diffusion. However, most drugs combined with albumin complexes are responsible for the drug’s transportation and distribution to the various compartment for cellular uptake [28]. The active metabolite of RSV also has the activity of anti-cancer and others (Figure 3). The study indicates that after 25 mg, oral administration of doses shows somewhere 70% absorption and peak plasma level of 491 ± 90 ng/mL, and a half-life of 9.2 ± 0.6 h. Most of the doses are found in the urine, which liquid chromatography-mass spectrometry (LC-MS) detects. RSV is easily uptaken by epithelial cells. Hence the concentration of drugs in these cells is high [29].

    Pharmacokinetic properties of RSV. CYP1A2: cytochrome P450 1A2; P-gp: P-glycoprotein; CYP2C9: cytochrome P450 2C9; CYP2D6: cytochrome P450 2D6; CYP3A4: cytochrome P450 3A4

    Topical route—A marked topical therapy requires excellent apparent permeability across the skin while showing less skin irritation. The topical application of RSV is limited because of its low permeability and poor skin retention [30]. This causes a decrease in the concentration of API required for efficacy, and less amount of drug reaches the target site. With molecular weight > 500 Da and a high partition coefficient, RSV fails to penetrate the stratum corneum. Furthermore, RSV is also associated with mild allergic reactions and erythema, which causes poor patient compliance. Hence, the lacks of effective delivery of RSV and undesirable skin interactions are the main reasons for its poor therapeutic efficacy.

    Parenteral route—Transition points, where pharmaceutical errors are more likely to occur, require specific attention for safe medication delivery. Compared to other drug delivery methods, parenteral administration is favored, such as in cases of cardiac arrest and anaphylactic shock [31]. This method of administration has many benefits, including avoiding first-pass metabolism, improved bioavailability, and consistent dose. Parenteral delivery, which is more controlled than oral administration in terms of dose and pace, results in more predictable pharmacodynamic and pharmacokinetic characteristics. As a result of the serious danger that could result from improper usage, several parenteral pharmaceuticals are categorized as high-alert drugs [32]. However, the parenteral administration of RSV causes severe adverse reactions like anaphylactic shock, muscle spasms, mild inflammation, etc. During multiple dosing, steady-state concentrations are unable to maintain leading to repetitive dosing leads to poor patient compliance.

    How RSV beneficial for cancer patients

    To show its effect, RSV exerts an effect on the various pathways, receptors, and other modulators. However, the causes of cancer are still unclear, but some hypothesis includes cytoplasmic cell division, abnormal cell, and gene hypothesis. RSV activates the SIRT1 [31], an enzyme located in the cell nucleus responsible for the deacetylation of histone and non-histone proteins, including transcription factors [32]. It is also required to regulate the various pathways that affect circadian rhythms, endothelial function, inflammation, immune function, metabolism, cell survival, and stress resistance. In cancer, SIRT1 regulates abnormal metabolic control, defects in the cell cycle, and inflammation [33]. It also regulates other conditions like cardiovascular diseases, obesity, and neurodegenerative diseases. RSV modulates the nuclear factor-κB (NF-κB) pathway by inhibiting the proteasome in human articular chondrocytes [34], which regulates the protein responsible for apoptosis and cell cycle progression. NF-κB regulates the antiapoptotic gene [35]. The active form of NF-κB results in the expression of a gene responsible for cell proliferating and protecting cells from apoptosis [programmed cell death (PCD)]. Whereas defects in the NF-κB result in increased apoptosis. NF-κB alters the activity of the caspase family enzyme, which is responsible for most apoptotic processes. RSV inhibits interleukin 1 (IL-1)-induced apoptosis simulation of caspase-3 and poly(ADP-ribose) polymerase (PARP) cleavage in human articular chondrocytes [36]. RSV suppressed the NF-κB regulated gene products such as matrix metalloproteinase-2 (MMP-2), MMP-3, MMP-9, vascular endothelial growth factor (VEGF), COX-2 and also inhibited apoptosis [B-cell lymphoma-extra large (Bcl-xL), B cell lymphoma protein-2 (Bcl-2), tumor necrosis factor-α (TNF-α) factor 1] [37].

    Suppression of MMP-2 lead to the inactivation of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) and p38 mitogen-activated protein kinases (MAPK) signaling pathway, which causes inhibition of A549 cell invasion beneficial for epithelial carcinoma. RSV inhibits the COX enzyme, which stops prostaglandin formation from arachidonic acid. Prostaglandin is responsible for synthesizing an inflammatory compound that promotes tumor cell proliferation or tumorigenesis. RSV inhibits the insulin-like growth factor-1 receptor (IGF-1R)/Akt/Wingless and int-1 (Wnt) signaling pathways that activate tumor protein (p53), leads to the suppression of colon cancer cell proliferation and induces apoptosis [38]. PI3K/Akt/mTOR pathway regulates various cellular activities such as cell proliferation and differentiation, cellular growth, survival, and mobility [39]. Components of these pathways show several abnormalities during various tumor growth, making it an exciting target for anti-cancer therapy. RSV inhibits PI3K/Akt/mTOR pathways, which are combined with other therapy [40]. RSV also downgraded cell cycle-regulated proteins such as retinoblastoma (Rb), cyclin-dependent kinase 2 (CDK2), CDK4, cyclin E, cyclin D1, and proliferating cell nuclear antigen (PCNA) [41], which inhibits the Akt pathways, causing the death of bladder cancer cell, liver cancer cell, and rats aortic vascular smooth muscle cell. RSV induces cell death, inhibits cell proliferation, and affects cell progression in the ovarian cancer cell line (A2780 cell) [42]. RSV induces the release of cytochrome c from the mitochondria to cytosol, which bind to apoptotic protease activating factor 1 (Apaf-1) to cause caspase activation and cause apoptosis or PCD. RSV also induces auto-phagocytosis, and the Bcl-2 and Bcl-xL do not inhibit the action of RSV in ovarian cancer cell A2780 [43].

    Nano-carrier: a critical appraisal

    A nanomaterial employed as a transport component for another chemical, such as a drug, is called a nanocarrier [44]. Micelles, polymers, carbon-based materials, liposomes, and other substances are frequently utilized as nanocarriers. Nanocarriers’ distinctive properties suggest a potential utility in chemotherapy, and they are now being investigated for their use in medication delivery [45]. Since micro-capillaries have a diameter of 200 nm and a diameter range of 1–1,000 nm for nanocarriers [46], nanomedicine frequently refers to objects with a diameter of less than 200 nm. Nanocarriers can deliver medications to parts of the body that would not typically be accessible due to their small size [47]. Because nanocarriers are so tiny, it is frequently challenging to administer substantial pharmacological doses using them. The low drug loading and drug encapsulation that frequently results from the emulsion procedures used to create nanocarriers presents a challenge for therapeutic application. Polymer conjugates, polymeric nanoparticles, lipid-based carriers, dendrimers, carbon nanotubes, and gold nanoparticles are a few of the newly identified nanocarriers [48]. Liposomes and micelles are two types of lipid-based carriers. Gold nanoshells and nanocages are two types of gold nanoparticles. Hydrophobic and hydrophilic medications can be distributed throughout the body thanks to the employment of several types of nanomaterial in nanocarriers. Since the human body is mainly made up of water, one important therapeutic advantage of nanocarriers is their capacity to transport hydrophobic medications to people successfully. Depending on the orientation of the phospholipid molecules, micelles can contain either hydrophilic or hydrophobic medicines [49]. Some nanocarriers have nanotube arrays that enable them to hold medications that are both hydrophilic and hydrophobic. Unwanted toxicity from the sort of nanomaterial being employed is one potential issue with nanocarriers. If inorganic nanomaterial builds up in specific cell organelles, it can also be hazardous to humans. To create safer, more efficient nanocarriers, new research is being done. Since protein-based nanocarriers are found in nature and often exhibit lower cytotoxicity than synthetic compounds, they hold promise for use as therapeutic agents [50, 51].

    Because they can deliver pharmaceuticals to site-specific targets, nanocarriers are helpful in the drug delivery process because they allow drugs to be administered in some organs or cells but not in others [52]. Site-specificity is a significant therapeutic advantage since it stops medications from being administered to the incorrect locations. Nanocarriers have promise for application in chemotherapy because they can lessen the chemotherapy’s harmful, widespread toxicity on the body’s healthy, rapidly dividing cells. Chemotherapy medications must be given to the tumor without spilling over into healthy tissue because they can be highly damaging to human cells.

    RSV nano-formulations

    Solid lipid nanoparticles

    Solid lipid matrices with surfactants display the best in vivo tolerability, good physico-chemical stability, and prolonged drug release. Solid lipid particles are made up of particles in the micrometer size range. RSV-loaded solid lipid nanoparticles (SLNs) show a better ability to suppress or inhibit the growth of human breast cancer cells (MDA-MB-231 cells) in comparison with free RSV [53]. Compared to liposomes, they have been described as lipidic drug carrier systems for topical applications that can replace polymers and enable large-scale manufacture at a relatively lower cost [54]. Solid lipid microparticles (SLMs) for RSV have the potential for oral delivery to improve their solubility and bioavailability, although having received less research attention than SLNs for skin applications. Several techniques, including solvent evaporation, melt dispersion, hot and cold homogenization, spray drying, and spray congealing, can be used to create SLMs [55]. D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) coated SLNs formulation of RSV given intravenously showed 9.37 folds higher plasma half-life and improved circulation time, better passive brain delivery of drug in Charles Foster rats [56].

    Microemulsions

    Because of their cutaneous tolerance and balanced hydrophilic-lipophilic character, microemulsions (MEs) are clear, colloidal, isotropic, and thermodynamically stable liquid dispersions of oil and water [57]. A multiphase system made up of water, oil, a surfactant, and a cosurfactant like alcohol that primarily serves as a co-solvent and demonstrates transparency by supplying globule size below 140 nm was the first to be referred to as a ME by Schulman in 1959 [58, 59]. A study shows that MEs gel-loaded RSV for sustained release in in vitro and ex vivo show 71.11 ± 0.47 and 68.15 ± 0.12 respectively in a 24 h period, which are tested for B16F10 melanoma cell lines [60]. MEs provide various benefits for topical distribution, including the capacity to dissolve lipophilic medicines effectively, improved skin permeability, and a longer release of both lipophilic and hydrophilic medications [57]. According to reports, oleic acid (OA) MEs exhibit a greater capacity for solubilizing RSV and a greater concentration of drug retention in the skin [61]. MEs have a high capacity for drug loading, and because they have a high capacity for solubilizing drugs [62], they can get through the stratum corneum barrier and partition the medication into the skin [63, 64]. A study indicated that MEs protect the RSV from ultraviolet B (UVB) radiation for up to 1 h and delay photodegradation which further helps in uptakes of RSV in the skin [65].

    Vesicular drug delivery systems

    Vesicular drug delivery systems consist of one or more concentric bilayers and are highly organized assemblies [6668]. They are created when amphiphilic building components self-assemble in the presence of water. These systems can localize the medication to the site of action, reducing the concentration of the drug at other places in the body, and making them effective for targeted drug delivery [69]. When compared to other non-vesiculized dose forms, this one help to achieve improved skin permeability and retention, as some of the popularly studied. Liposomes, transferosomes, ethosomes, and niosomes are examples of vesicular systems for topical delivery [70, 71].

    Liposomes

    An aqueous core is encircled by a hydrophobic lipid bilayer membrane that contains phospholipids and cholesterol in liposomes [72], which are biocompatible and biodegradable vesicles. They allow for better active ingredient absorption through the skin. Although they are simple to prepare, they are prone to structural failure and oxidation. The design, content, size, and drug-release properties of liposomes are flexible [73, 74]. According to published clinical research, liposomal anticancer medication has less toxicity and better tolerance [75]. For in vitro skin permeation and in vivo antineoplastic effect [76], for example, in combination with RSV and 5-fluorouracil (5-FU) in a liposome shows better skin cancer activity as compared to a single drug [77]. A study conducted by researchers concluded that RSV-loaded liposomes show stable, good loading efficiency (70–75%), and prolonged release in vitro which improved the activity of anti-proliferation and apoptosis in U-87 MG cell line and xenograft-bearing mice [78]. Another study shows that 4-Carboxybutyl triphenylphosphonium bromide (TPP)-based liposomes showed better mitochondrial targeting and enhance the activity of RSV in B16F10 cell lines [79].

    Transferosomes

    Transferosomes are extremely flexible, self-assembled, and ultradeformable vesicles with an aqueous core and a complex lipid bilayer on each side [80]. These vesicles are self-regulating and self-optimizing because of their structure and composition. Transferosomes can spontaneously penetrate the stratum corneum (SC) because they can efficiently cross a variety of transport barriers [81]. They have better efficacy in sustained release applications for topical medication administration and are more elastic than liposomes [82, 83]. A study showed that transferosomes encapsulated RSV has more penetration power (27.5%) in comparison with free RSV [84]. Particle size and shape, zeta potential, viscosity, entrapment effectiveness, deformability, in vitro drug release, kinetics, and drug retention were characterization characteristics. The transferosome was created using the solvent evaporation method, whereas the reverse-phase evaporation method was employed to create the liposomes and niosomes.

    Niosomes

    Niosomes are composed of nonionic surfactant vesicles and are similar to liposomes in structure. These formulations are becoming more and more significant for cutaneous drug administration because they have traits such as improved drug penetration, prolonged drug release, increased drug stability, and the capacity to transport both hydrophilic and lipophilic drugs [85]. Pando et al. [86] formulated a niosomes taking surfactant G64, OA, and linoleic acid (LA) as a penetration enhancer and loaded with RSV which was then evaluated. Results are very interesting, the niosomes prepared by ethanol injection modified method (EIM) show a 21% increase in penetration enhancement, ex vivo experiment held in Franz diffusion cells collected from newborn pig skin [86]. Different nano-formulations loaded with RSV improving therapeutic activity are listed in Table 1.

    A 10-year literature review of different nano-formulations of RSV

    Route of administrationDrug (API)ExcipientFormulationObservationsReference
    Parenteral delivery (intravenous)trans-RSVTPGSSLNShowed 9.37 folds higher plasma half-life and improved circulation time, better passive brain delivery of drug in Charles Foster rats.[56]
    Oral deliveryRSVMagnesium dihydroxideSolid dispersionImproved solubility and bioavailability (3.3-fold) as compared to RSV alone.[87]
    A mixed lipid phase (castor oil/Capmul MCM 1:1) and a mixed surfactant phase (Kolliphor EL/Kolliphor RH 40 1:1)Self-micro emulsifying drug delivery system (SMEDDS)Formulation improved the bioavailability and reduce toxicity which is further used in the supplement as well as the pharmaceutical industry.[88]
    poly (DL-lactide-co-glycolide) (PLGA)Systematically optimized nanoparticlesKa and AUC are 7.17 and 10.6 folds respectively while bioavailability showed 2.78 folds.[89]
    Phospholipids, Sylysia 350, TPGSSolidified phospholipid-TPGSIncreased bioavailability by dissolution rate and absorption.[90]
    ZeinNano capsulationImproved bioavailability and permeability of 1.15 folds.[91]
    Di stearoyl phosphatidyl choline, sodium taurocholate, cholesterolProliposomal formulationAUC and Cmax increased by 2 folds.[92]
    Nasal deliveryRSV8.75% w/v chitosan solution, SLMs, chitosanLMs uncoated or coated with chitosanImprove Cmax (60 min) about 9.7 ± 1.9 μg/mL.
    Effective targeting of brain drug delivery by nasal route.
    [93]
    TransferosomesIntranasal transferosome mucoadhesive gelImproved Cmax and AUC by 2.15 and 22.5 folds respectively as compared to RSV suspension alone.[94]
    Topical deliveryRSVGelot 64 (surfactant), OAs and LAsNiosomesTopical penetration increased up to 21%.[86]
    DendrimerDendrimer nanotechnology78.06% of the formulation showed intradermal permeation and only 37.33% RSV alone.[13]
    EugenolNano emulsionThe highest RSV penetration into the stratum corneum is about 9.55 (ratio compared to saturated RSV).[95]
    LimoneneNano emulsionThe highest permeation of drug to the skin is about 12.61 (ratio compared to saturated RSV).[95]
    Display full size

    AUC: area under the curve; Cmax: peak plasma concentration; LMs: lipid microparticles

    Patent survey of RSV nanoformulation

    However, many patents have been filled in the last two decades, which shows a significant interest in this area. Many challenges have been addressed, and various methods have continuously improved product quality by innovation in formulating RSV. Some of them are nanoformulation which improves severe problem that occurs during the drug delivery of RSV. A list of such patented formulations and methods is given in Table 2.

    Patent survey of different nano-formulations of RSV

    Patent numberInventor nameInventionDetailsReference
    WO-2012017451-A1Pratibha Omray, Vinay Kumar TripathiA bio-stabilized resveratrol formulationA herbal medicament contains 50–80% RSV and other ingredients which helps to prevent and treat cancer, Alzheimer’s disease, obesity, and others.[96]
    US20110281957A1Eric KuhrtsEnhanced bioactive formulations of resveratrolEnhanced solubility of RSV which is used for the treatment of disease states like cancer.[97]
    ES2798403T3Dariush Behnam, Marshall A HaywardResveratrol solubilization product for pharmaceutical purposesRSV solubilization product is used for pharmaceutical purposes and helps increase plasma level for therapeutics effects and also reduce GIT side effects as compared with natural RSV.[98]
    WO2001030336A2John M. Pezzuto, Richard C. Moon, Mei-Shiang Jang, Aomar Ouali, Shengzhao Lin, Karla Slowing BarillasPharmaceutical formulations comprising resveratrol and use thereofDesigned the formulation for topical delivery which are helpful in preventing and treating the skin disorder caused by inflammation, sunburn, and aging.[99]
    WO2010059628A1Arthur S. Polans, Lalita Subramanian, Ronak Vakil, Glen S. KwonWater-soluble formulations of resveratrol and uses thereofFormulation of RSV with solubilizer poloxamer 334 and the solution can be used for the medicament of cancer.[100]
    CN104688715BWang Zhicheng, Zhu Kexin, Wang Bing, Cai Feng, Ren Jianlin, Zhang Tong, Zhang Qi, Zong ShiyuA kind of resveratrol solid lipid nano granule and preparation method thereofThis invention helps to reduce particle diameter, drug loading and bioavailability is high and has fast absorption, and is convenient to take.[101]
    CN105126116BXiao Chunsheng, Chen Xin, Ding Jianxun, Zhuang Xiuli, Chen XuesiA kind of resveratrol nano particle and preparation method thereofThe formation of nanoparticle cross-linked structure with RSV increased water solubility and improved stability.[102]
    CN101214225AOuyang Wuqing, Yang BaopingResveratrol nano emulsion anti-cancer medicineRSV nano emulsion increase half-life and stability and also prevent RSV to get oxygenated.[103]
    Display full size

    Future challenges

    There is a number of RSV studies that are listed on ClinicalTrials.gov, the database of publicly and privately financed human clinical research. Numerous of these trials have been conducted to assess RSV’s pharmacokinetics, bioavailability, safety, and tolerability. Only a few of the studies mentioned are concerned with determining whether RSV is effective in treating particular malignancies [104]. Trans-RSV, Polygonum cuspidatum (Japanese knotweed) extract, SRT501 (micronized RSV), RSV-rich seedless red grapes/grape juices (muscadine grapes), and micro-encapsulated RSV are some of the types of RSV used in these studies. Numerous cancer types, including multiple myeloma, breast cancer, follicular lymphoma, and neuroendo-crine tumors are the subject of these trials, although the majority of them examine how RSV affects the growth of colon malignancies. In reality, RSV has shown to be slightly effective in colon cancer clinical trials, which may be related to RSV’s potential direct contact and extended exposure to colonic tissues [105]. The gut epithelium also appears to be well-suited for absorbing nutrients and active chemicals from food and food components [106].

    According to ClinicalTrials.gov, it is exciting to note that while planning the new clinical trials, researchers used what they had learned from earlier trials, which should provide improved outcomes. Two of the four ongoing trials are aimed at gastrointestinal malignancies. The goal of one of these trials (NCT00433576) is to determine the ideal RSV dosage that will produce bioactive levels in the colon mucosa. This is significant because an ideal dose has not yet been identified despite extensive RSV studies on colon cancer [106]. Additionally, the researchers will determine whether there are correlated levels of COX-2 and pyrimido[1,2-α]purin-10(3H)-one (M1G) adduct in cancer tissues to examine the mechanistic consequences of RSV treatment of colorectal adenocarcinoma. Given that both of these molecules have been proven to be regulated in colon cancer, this might offer some relevant information. The goal of the other ongoing colon cancer experiment (NCT00578396), which uses seedless red grapes, is to ascertain the highest dietary RSV levels possible. This may be crucial for colon cancer prevention because it would be simple to develop new dietary guidelines for the general public. These trials ought to yield some information that will be beneficial in more in-depth human research that aims to make RSV available in clinics for the treatment of diseases. Diets high in RSV may also be encouraged for improved health and illness prevention [107, 108].

    Conclusions

    The vast range of applicability of RSV makes it a center of attraction for the investigation and search for the novelty to improve pharmacokinetics such as (solubility and bioavailability) and toxicological effects on humans. Several nanoformulation such as niosomes, liposomes, MEs, SLNs, nanocapsules, and other formulations enhanced the therapeutic efficacy and solubility of RSV, also improving the bioavailability of oral and increasing retention and penetration of topical formulation many folds, which are discussed in Table 1. Although RSV has many beneficial effects on the body most highlighted are anti-oxidant and anti-cancer activity. It regulates various pathway inhibition (PI3K/Akt/mTOR, IGF-1R/Akt/Wnt, IL-1 induced apoptosis simulation), modulation (NF-κB), and activation (SIRT1) and induction (release of cytochrome c, auto phagocytosis) to exert its effect. Hence, some patent formulations could be a sign of interest. Nevertheless, apart from anti-cancer activity, its applicability is not limited due to its uses in nutraceuticals, herbal and pharmaceutical industries.

    Abbreviations

    AKT:

    protein kinase B

    API:

    active pharmaceutical ingredient

    COXs:

    cyclooxygenases

    MEs:

    microemulsions

    MMP-2:

    matrix metalloproteinase-2

    mTOR:

    mammalian target of rapamycin

    NF-κB:

    nuclear factor-κB

    OA:

    oleic acid

    PI3K:

    phosphoinositol 3 kinase

    RSV:

    resveratrol

    SIRTs:

    sirtuins

    SLMs:

    solid lipid microparticles

    SLNs:

    solid lipid nanoparticles

    TPGS:

    D-α-tocopheryl polyethylene glycol 1000 succinate

    Declarations

    Acknowledgments

    The authors are highly thankful to ISF College of Pharmacy, Moga for providing the necessary facilities for carrying out the review work.

    Author contributions

    AK and DS conceptualized the idea and prepared the first draft of the article. BDK and AS finalized the draft and approved it for final submission. DS submit the first draft of the article to the journal.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2022.

    References

    The genetics of cancer [Internet]. NCI; c2022 [cited 2022 Jul 17]. Available from: https://www.cancer.gov/about-cancer/causes-prevention/genetics
    Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:733. [DOI] [PubMed]
    Cancer statistics [Internet]. NCI; c2022 [cited 2022 Jul 25]. Available from: https://www.cancer.gov/about-cancer/understanding/statistics
    George P. Concerns regarding the safety and toxicity of medicinal plants. J Appl Pharm Sci. 2011;6:404.
    Pezzuto JM. Resveratrol: twenty years of growth, development and controversy. Biomol Ther (Seoul). 2019;27:114. [DOI] [PubMed] [PMC]
    Springer M, Moco S. Resveratrol and its human metabolites-effects on metabolic health and obesity. Nutrients. 2019;11:143. [DOI] [PubMed] [PMC]
    Singh CK, George J, Ahmad N. Resveratrol-based combinatorial strategies for cancer management. Ann N Y Acad Sci. 2013;1290:11321. [DOI] [PubMed] [PMC]
    Din FU, Aman W, Ullah I, Qureshi OS, Mustapha O, Shafique S, et al. Effective use of nanocarriers as drug delivery systems for the treatment of selected tumors. Int J Nanomedicine. 2017;12:7291309. [DOI] [PubMed] [PMC]
    Khalid A, Persano S, Shen H, Zhao Y, Blanco E, Ferrari M, et al. Strategies for improving drug delivery: nanocarriers and microenvironmental priming. Expert Opin Drug Deliv. 2017;14:86577. [DOI] [PubMed] [PMC]
    Galiniak S, Aebisher D, Bartusik-Aebisher D. Health benefits of resveratrol administration. Acta Biochim Pol. 2019;66:1321. [DOI] [PubMed]
    Wang H, Yang YJ, Qian HY, Zhang Q, Xu H, Li JJ. Resveratrol in cardiovascular disease: what is known from current research? Heart Fail Rev. 2012;17:43748. [DOI] [PubMed]
    Moran BW, Anderson FP, Devery A, Cloonan S, Butler WE, Varughese S, et al. Synthesis, structural characterisation and biological evaluation of fluorinated analogues of resveratrol. Bioorg Med Chem. 2009;17:451022. [DOI] [PubMed]
    Pentek T, Newenhouse E, O’Brien B, Chauhan AS. Development of a topical resveratrol formulation for commercial applications using dendrimer nanotechnology. Molecules. 2017;22:137. [DOI] [PubMed] [PMC]
    Li M, Kildegaard KR, Chen Y, Rodriguez A, Borodina I, Nielsen J. De novo production of resveratrol from glucose or ethanol by engineered Saccharomyces cerevisiae. Metab Eng. 2015;32:111. [DOI] [PubMed]
    Chang X, Heene E, Qiao F, Nick P. The phytoalexin resveratrol regulates the initiation of hypersensitive cell death in Vitis cell. PLoS One. 2011;6:e26405. [DOI] [PubMed] [PMC]
    Gu J, Li Z, Chen H, Xu X, Li Y, Gui Y. Neuroprotective effect of trans-resveratrol in mild to moderate Alzheimer disease: a randomized, double-blind trial. Neurol Ther. 2021;10:90517. [DOI] [PubMed] [PMC]
    Shaito A, Posadino AM, Younes N, Hasan H, Halabi S, Alhababi D, et al. Potential adverse effects of resveratrol: a literature review. Int J Mol Sci. 2020;21:2084. [DOI] [PubMed] [PMC]
    Robinson K, Mock C, Liang D. Pre-formulation studies of resveratrol. Drug Dev Ind Pharm. 2015;41:14649. [DOI] [PubMed] [PMC]
    Meng T, Xiao D, Muhammed A, Deng J, Chen L, He J. Anti-inflammatory action and mechanisms of resveratrol. Molecules. 2021;26:229. [DOI] [PubMed] [PMC]
    Liu Q, Guan J, Sun Z, Shen X, Li L, Jin L, et al. Influence of stabilizer type and concentration on the lung deposition and retention of resveratrol nanosuspension-in-microparticles. Int J Pharm. 2019;569:118562. [DOI] [PubMed]
    de Queiroz KB, Dos Santos Fontes Pereira T, Araújo MSS, Gomez RS, Coimbra RS. Resveratrol acts anti-inflammatory and neuroprotective in an infant rat model of pneumococcal meningitis by modulating the hippocampal miRNome. Mol Neurobiol. 2018;55:886984. [DOI] [PubMed]
    Ren B, Kwah MX, Liu C, Ma Z, Shanmugam MK, Ding L, et al. Resveratrol for cancer therapy: challenges and future perspectives. Cancer Lett. 2021;515:6372. [DOI] [PubMed]
    Walle T. Bioavailability of resveratrol. Ann N Y Acad Sci. 2011;1215:915. [DOI] [PubMed]
    Annaji M, Poudel I, Boddu SHS, Arnold RD, Tiwari AK, Babu RJ. Resveratrol-loaded nanomedicines for cancer applications. Cancer Rep (Hoboken). 2021;4:e1353. [DOI] [PubMed] [PMC]
    Tomé-Carneiro J, Larrosa M, González-Sarrías A, Tomás-Barberán FA, García-Conesa MT, Espín JC. Resveratrol and clinical trials: the crossroad from in vitro studies to human evidence. Curr Pharm Des. 2013;19:606493. [DOI] [PubMed] [PMC]
    Sharifi-Rad J, Quispe C, Alfred MA, Anil N, Lombardi N, Cinquanta L, et al. Current trends on resveratrol bioactivities to treat periodontitis. Food Biosci. 2021;42:101205. [DOI]
    Ndiaye F, Vuong T, Duarte J, Aluko RE, Matar C. Anti-oxidant, anti-inflammatory and immunomodulating properties of an enzymatic protein hydrolysate from yellow field pea seeds. Eur J Nutr. 2012;51:2937. [DOI] [PubMed]
    Lançon A, Delmas D, Osman H, Thénot JP, Jannin B, Latruffe N. Human hepatic cell uptake of resveratrol: involvement of both passive diffusion and carrier-mediated process. Biochem Biophys Res Commun. 2004;316:11327. [DOI] [PubMed]
    Walle T, Hsieh F, DeLegge MH, Oatis JE Jr, Walle UK. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos. 2004;32:137782. [DOI] [PubMed]
    Sharma B, Iqbal B, Kumar S, Ali J, Baboota S. Resveratrol-loaded nanoemulsion gel system to ameliorate UV-induced oxidative skin damage: from in vitro to in vivo investigation of antioxidant activity enhancement. Arch Dermatol Res. 2019;311:77393. [DOI] [PubMed]
    Zhao M, Ko SY, Garrett IR, Mundy GR, Gutierrez GE, Edwards JR. The polyphenol resveratrol promotes skeletal growth in mice through a sirtuin 1-bone morphogenic protein 2 longevity axis. Br J Pharmacol. 2018;175:418392. [DOI] [PubMed] [PMC]
    Thiel G, Rössler OG. Resveratrol regulates gene transcription via activation of stimulus-responsive transcription factors. Pharmacol Res. 2017;117:16676. [DOI] [PubMed]
    Chalkiadaki A, Guarente L. The multifaceted functions of sirtuins in cancer. Nat Rev Cancer. 2015;15:60824. [DOI] [PubMed]
    Haseeb A, Chen D, Haqqi TM. Delphinidin inhibits IL-1β-induced activation of NF-κB by modulating the phosphorylation of IRAK-1Ser376 in human articular chondrocytes. Rheumatology (Oxford). 2013;52:9981008. [DOI] [PubMed] [PMC]
    Vallianou NG, Evangelopoulos AA, Geladari EV, Kazazis CE. Resveratrol and cancer. Hosp Chron. 2015;10:13744. [DOI]
    Shakibaei M, John T, Seifarth C, Mobasheri A. Resveratrol inhibits IL-1β-induced stimulation of caspase-3 and cleavage of PARP in human articular chondrocytes in vitro. Ann N Y Acad Sci. 2007;1095:55463. [DOI] [PubMed]
    Csaki C, Mobasheri A, Shakibaei M. Synergistic chondroprotective effects of curcumin and resveratrol in human articular chondrocytes: inhibition of IL-1β-induced NF-κB-mediated inflammation and apoptosis. Arthritis Res Ther. 2009;11:R165. [DOI] [PubMed] [PMC]
    Vanamala J, Reddivari L, Radhakrishnan S, Tarver C. Resveratrol suppresses IGF-1 induced human colon cancer cell proliferation and elevates apoptosis via suppression of IGF-1R/Wnt and activation of p53 signaling pathways. BMC Cancer. 2010;10:238. [DOI] [PubMed] [PMC]
    Xu W, Yang Z, Lu N. A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adh Migr. 2015;9:31724. [DOI] [PubMed] [PMC]
    Morgensztern D, McLeod HL. PI3K/Akt/mTOR pathway as a target for cancer therapy. Anticancer Drugs. 2005;16:797803. [DOI] [PubMed]
    Kim YA, Lee WH, Choi TH, Rhee SH, Park KY, Choi YH. Involvement of p21WAF1/CIP1, pRB, Bax and NF-kappaB in induction of growth arrest and apoptosis by resveratrol in human lung carcinoma A549 cells. Int J Oncol. 2003;23:11439. [DOI] [PubMed]
    Liu Y, Tong L, Luo Y, Li X, Chen G, Wang Y. Resveratrol inhibits the proliferation and induces the apoptosis in ovarian cancer cells via inhibiting glycolysis and targeting AMPK/mTOR signaling pathway. J Cell Biochem. 2018;119:616272. [DOI] [PubMed]
    Opipari AW Jr, Tan L, Boitano AE, Sorenson DR, Aurora A, Liu JR. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res. 2004;64:696703. [DOI] [PubMed]
    Kumari P, Ghosh B, Biswas S. Nanocarriers for cancer-targeted drug delivery. J Drug Target. 2016;24:17991. [DOI] [PubMed]
    Swathi G, Prasanthi NL, Manikiran SS, Ramarao N. Solid lipid nanoparticles: colloidal carrier systems for drug delivery. ChemInform. 2012;43. [DOI]
    Danaei M, Dehghankhold M, Ataei S, Hasanzadeh Davarani F, Javanmard R, Dokhani A, et al. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics. 2018;10:57. [DOI] [PubMed] [PMC]
    Rizvi SAA, Saleh AM. Applications of nanoparticle systems in drug delivery technology. Saudi Pharm J. 2018;26:6470. [DOI] [PubMed] [PMC]
    Bolhassani A, Javanzad S, Saleh T, Hashemi M, Aghasadeghi MR, Sadat SM. Polymeric nanoparticles: potent vectors for vaccine delivery targeting cancer and infectious diseases. Hum Vaccin Immunother. 2014;10:32132. [DOI] [PubMed] [PMC]
    Joseph M, Trinh HM, Mitra AK. Chapter 7 - Peptide and protein-based therapeutic agents*. In: Mitra AK, Cholkar K, Mandal A, editors. Emerging nanotechnologies for diagnostics, drug delivery and medical devices. Boston: Elsevier; 2017. pp. 14567. [DOI]
    Verma D, Gulati N, Kaul S, Mukherjee S, Nagaich U. Protein based nanostructures for drug delivery. J Pharm (Cairo). 2018;2018:9285854. [DOI] [PubMed] [PMC]
    Yin L, Yuvienco C, Montclare JK. Protein based therapeutic delivery agents: contemporary developments and challenges. Biomaterials. 2017;134:91116. [DOI] [PubMed] [PMC]
    Girdhar V, Patil S, Banerjee S, Singhvi G. Nanocarriers for drug delivery: mini review. Curr Nanomed. 2018;8:8899. [DOI]
    Wang W, Zhang L, Chen T, Guo W, Bao X, Wang D, et al. Anticancer effects of resveratrol-loaded solid lipid nanoparticles on human breast cancer cells. Molecules. 2017;22:1814. [DOI] [PubMed] [PMC]
    Ravikumar P, Katariya M, Patil S, Tatke P, Pillai R. Skin delivery of resveratrol encapsulated lipidic formulation for melanoma chemoprevention. J Microencapsul. 2019;36:53551. [DOI] [PubMed]
    Passerini N, Qi S, Albertini B, Grassi M, Rodriguez L, Craig DQ. Solid lipid microparticles produced by spray congealing: influence of the atomizer on microparticle characteristics and mathematical modeling of the drug release. J Pharm Sci. 2010;99:91631. [DOI] [PubMed]
    Vijayakumar MR, Kumari L, Patel KK, Vuddanda PR, Vajanthri KY, Mahtoc SK, et al. Intravenous administration of trans-resveratrol-loaded TPGS-coated solid lipid nanoparticles for prolonged systemic circulation, passive brain targeting and improved in vitro cytotoxicity against C6 glioma cell lines. RSC Adv. 2016;6:5033648. [DOI]
    Shaker DS, Ishak RAH, Ghoneim A, Elhuoni MA. Nanoemulsion: a review on mechanisms for the transdermal delivery of hydrophobic and hydrophilic drugs. Sci Pharm. 2019;87:17. [DOI]
    Srikanth K, Gupta VR, Manvi SR, Devanna N. Particulate carrier systems: a review. IRJP. 2012;3:226.
    Malik MA, Wani MY, Hashim MA. Microemulsion method: a novel route to synthesize organic and inorganic nanomaterials: 1st nano update. Arabian J Chem. 2012;5:397417. [DOI]
    Ravikumar P, Tatke P. Design of an encapsulated topical formulation for chemoprevention of skin cancer. Int J Pharm Sci Res. 2019;10:30919. [DOI]
    Souto EB, Cano A, Martins-Gomes C, Coutinho TE, Zielińska A, Silva AM. Microemulsions and nanoemulsions in skin drug delivery. Bioengineering (Basel). 2022;9:158. [DOI] [PubMed] [PMC]
    Patil NH, Devarajan PV. Chapter 20 - Colloidal carriers for noninvasive delivery of insulin. In: Ohshima H, Makino K, editors. Colloid and interface science in pharmaceutical research and development. Amsterdam: Elsevier; 2014. pp. 41142. [DOI]
    Hathout RM, Mansour S, Mortada ND, Geneidi AS, Guy RH. Uptake of microemulsion components into the stratum corneum and their molecular effects on skin barrier function. Mol Pharm. 2010;7:126673. [DOI] [PubMed]
    Heuschkel S, Goebel A, Neubert RH. Microemulsions--modern colloidal carrier for dermal and transdermal drug delivery. J Pharm Sci. 2008;97:60331. [DOI] [PubMed]
    Juškaitė V, Ramanauskienė K, Briedis V. Testing of resveratrol microemulsion photostability and protective effect against UV induced oxidative stress. Acta Pharm. 2017;67:24756. [DOI] [PubMed]
    Biju S, Talegaonkar S, Mishra P, Khar R. Vesicular systems: an overview. Indian J Pharm Sci. 2006;68:14153. [DOI]
    Pandita A, Sharma P. Pharmacosomes: an emerging novel vesicular drug delivery system for poorly soluble synthetic and herbal drugs. ISRN Pharm. 2013;2013:348186. [DOI] [PubMed] [PMC]
    Witika BA, Mweetwa LL, Tshiamo KO, Edler K, Matafwali SK, Ntemi PV, et al. Vesicular drug delivery for the treatment of topical disorders: current and future perspectives. J Pharm Pharmacol. 2021;73:142741. [DOI] [PubMed]
    Jain S, Jain V, Mahajan SC. Lipid based vesicular drug delivery systems. Adv Pharm. 2014;2014:112. [DOI]
    Cevc G. Transfersomes, liposomes and other lipid suspensions on the skin: permeation enhancement, vesicle penetration, and transdermal drug delivery. Crit Rev Ther Drug Carrier Syst. 1996;13:257388. [DOI] [PubMed]
    Singh D, Pradhan M, Nag M, Singh MR. Vesicular system: versatile carrier for transdermal delivery of bioactives. Artif Cells Nanomed Biotechnol. 2015;43:28290. [DOI] [PubMed]
    Nakhaei P, Margiana R, Bokov DO, Abdelbasset WK, Jadidi Kouhbanani MA, Varma RS, et al. Liposomes: structure, biomedical applications, and stability parameters with emphasis on cholesterol. Front Bioeng Biotechnol. 2021;9:705886. [DOI] [PubMed] [PMC]
    Soni V, Chandel S, Jain P, Asati S. Chapter 5 - Role of liposomal drug-delivery system in cosmetics. In: Grumezescu AM, editor. Nanobiomaterials in galenic formulations and cosmetics. William Andrew Publishing; 2016. pp. 93120. [DOI]
    Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine. 2006;1:297315. [PubMed] [PMC]
    Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286. [DOI] [PubMed] [PMC]
    Zeisig R, Arndt D, Stahn R, Fichtner I. Physical properties and pharmacological activity in vitro and in vivo of optimised liposomes prepared from a new cancerostatic alkylphospholipid. Biochim Biophys Acta. 1998;1414:23848. [DOI] [PubMed]
    Cosco D, Paolino D, Maiuolo J, Marzio LD, Carafa M, Ventura CA, et al. Ultradeformable liposomes as multidrug carrier of resveratrol and 5-fluorouracil for their topical delivery. Int J Pharm. 2015;489:110. [DOI] [PubMed]
    Jhaveri A, Deshpande P, Pattni B, Torchilin V. Transferrin-targeted, resveratrol-loaded liposomes for the treatment of glioblastoma. J Control Release. 2018;277:89101. [DOI] [PubMed] [PMC]
    Kang JH, Ko YT. Enhanced subcellular trafficking of resveratrol using mitochondriotropic liposomes in cancer cells. Pharmaceutics. 2019;11:423. [DOI] [PubMed] [PMC]
    Narvekar MNG, Redkar MMR, Bhosale MNS. Self-assembled ultradeformable phospholipid vesicles with edge activators for delivery of transcutaneous bioactives. Indo Am J Pharm Res. 2019;9:54352. [DOI]
    Opatha SAT, Titapiwatanakun V, Chutoprapat R. Transfersomes: a promising nanoencapsulation technique for transdermal drug delivery. Pharmaceutics. 2020;12:855. [DOI] [PubMed] [PMC]
    Das B, Nayak AK, Mallick S. Chapter 7 - Transferosomes: a novel nanovesicular approach for drug delivery. In: Nayak AK, Hasnain MS, Aminabhavi TM, Torchilin VP, editors. Systems of nanovesicular drug delivery. Academic Press; 2022. pp. 10314. [DOI]
    Kumar PK, Kumar RS. Review on transferosomes and transferosomal gels. J Pharm Res Int. 2021;33:11426. [DOI]
    Wu PS, Li YS, Kuo YC, Tsai SJ, Lin CC. Preparation and evaluation of novel transfersomes combined with the natural antioxidant resveratrol. Molecules. 2019;24:600. [DOI] [PubMed] [PMC]
    Hamishehkar H, Rahimpour Y, Kouhsoltani M. Niosomes as a propitious carrier for topical drug delivery. Expert Opin Drug Deliv. 2013;10:26172. [DOI] [PubMed]
    Pando D, Matos M, Gutiérrez G, Pazos C. Formulation of resveratrol entrapped niosomes for topical use. Colloids Surf B Biointerfaces. 2015;128:398404. [DOI] [PubMed]
    Spogli R, Bastianini M, Ragonese F, Iannitti RG, Monarca L, Bastioli F, et al. Solid dispersion of resveratrol supported on magnesium diHydroxide (Resv@MDH) microparticles improves oral bioavailability. Nutrients. 2018;10:1925. [DOI] [PubMed] [PMC]
    Seljak KB, Berginc K, Trontelj J, Zvonar A, Kristl A, Gašperlin M. A self-microemulsifying drug delivery system to overcome intestinal resveratrol toxicity and presystemic metabolism. J Pharm Sci. 2014;103:3491500. [DOI] [PubMed]
    Singh G, Pai RS. Optimized PLGA nanoparticle platform for orally dosed trans-resveratrol with enhanced bioavailability potential. Expert Opin Drug Deliv. 2014;11:64759. [DOI] [PubMed]
    Liu C, Tong P, Yang R, You Y, Liu H, Zhang T. Solidified phospholipid-TPGS as an effective oral delivery system for improving the bioavailability of resveratrol. J Drug Delivery Sci Technol. 2019;52:76977. [DOI]
    Jayan H, Leena MM, Sundari SKS, Moses JA, Anandharamakrishnan C. Improvement of bioavailability for resveratrol through encapsulation in zein using electrospraying technique. J Funct Foods. 2019;57:41724. [DOI]
    Basavaraj S, Betageri GV. Improved oral delivery of resveratrol using proliposomal formulation: investigation of various factors contributing to prolonged absorption of unmetabolized resveratrol. Expert Opin Drug Deliv. 2014;11:493503. [DOI] [PubMed]
    Trotta V, Pavan B, Ferraro L, Beggiato S, Traini D, Des Reis LG, et al. Brain targeting of resveratrol by nasal administration of chitosan-coated lipid microparticles. Eur J Pharm Biopharm. 2018;127:2509. [DOI] [PubMed]
    Salem HF, Kharshoum RM, Abou-Taleb HA, Naguib DM. Nanosized transferosome-based intranasal in situ gel for brain targeting of resveratrol: formulation, optimization, in vitro evaluation, and in vivo pharmacokinetic study. AAPS PharmSciTech. 2019;20:181. [DOI] [PubMed]
    Nastiti CMRR, Ponto T, Mohammed Y, Roberts MS, Benson HAE. Novel nanocarriers for targeted topical skin delivery of the antioxidant resveratrol. Pharmaceutics. 2020;12:108. [DOI] [PubMed] [PMC]
    Omray P, Tripathi VK, inventors; Khandelwal S, assignee. A bio-stabilized resveratrol formulation. WO-2012017451-A1. 2010 Aug 3.
    Kuhrts E, inventor; Lipoprotein Technologies Inc., assignee. Enhanced bioactive formulations of resveratrol. US2011281957A1. 2011 Nov 17.
    Behnam D, Hayward MA, inventors; Aquanova AG, assignee. Resveratrol solubilization product for pharmaceutical purposes. ES2798403T3. 2020 Dec 11.
    Pezzuto JM, Moon RC, Jang MS, Ouali A, Lin S, Barillas KS, inventors. Pharmaceutical formulations comprising resveratrol and use thereof. WO2001030336A2. 2001 May 3.
    Polans AS, Subramaning L, Vakil R, Kwon GS, inventors. Water-soluble formulations of resveratrol and uses thereof. WO2010059628A1. 2010 May 27.
    Wang Z, Zhu K, Wang B, Cai F, Ren J, Zhang T, et al., inventors; Shanghai traditional chinese medicine hospital, assignee. A kind of resveratrol solid lipid nano granule and preparation method thereof. CN104688715B. 2017 Oct 10.
    Xiao C, Chen X, Ding J, Zhuang X, Chen X, inventors. A kind of resveratrol nano particle and preparation method thereof. CN105126116B. 2015 Dec 9.
    Ouyang W, Yang B, inventors. Resveratrol nano emulsion anti-cancer medicine. CN101214225A. 2008 Jul 9.
    Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, et al. The role of resveratrol in cancer therapy. Int J Mol Sci. 2017;18:2589. [DOI] [PubMed] [PMC]
    Talib WH, Alsayed AR, Farhan F, Al Kury LT. Resveratrol and tumor microenvironment: mechanistic basis and therapeutic targets. Molecules. 2020;25:4282. [DOI] [PubMed] [PMC]
    Singh CK, Ndiaye MA, Ahmad N. Resveratrol and cancer: challenges for clinical translation. Biochim Biophys Acta. 2015;1852:117885. [DOI] [PubMed] [PMC]
    Markus MA, Morris BJ. Resveratrol in prevention and treatment of common clinical conditions of aging. Clin Interv Aging. 2008;3:3319. [DOI] [PubMed] [PMC]
    Meng X, Zhou J, Zhao CN, Gan RY, Li HB. Health benefits and molecular mechanisms of resveratrol: a narrative review. Foods. 2020;9:340. [DOI] [PubMed] [PMC]